Next Article in Journal
Isolation and Characterization of a Cholesterol-Lowering Bacteria from Bubalus bubalis Raw Milk
Next Article in Special Issue
Probiotic and Antifungal Attributes of Lactic Acid Bacteria Isolates from Naturally Fermented Brazilian Table Olives
Previous Article in Journal
Recent Developments in Lignocellulosic Biofuels, a Renewable Source of Bioenergy
Previous Article in Special Issue
Fermentation of Cereals and Legumes: Impact on Nutritional Constituents and Nutrient Bioavailability
 
 
Font Type:
Arial Georgia Verdana
Font Size:
Aa Aa Aa
Line Spacing:
Column Width:
Background:
Review

Fermented Foods and Their Role in Respiratory Health: A Mini-Review

by
Periyanaina Kesika
1,2,
Subramanian Thangaleela
1,
Bhagavathi Sundaram Sivamaruthi
2,*,
Muruganantham Bharathi
1 and
Chaiyavat Chaiyasut
1,*
1
Innovation Center for Holistic Health, Nutraceuticals and Cosmeceuticals, Faculty of Pharmacy, Chiang Mai University, Chiang Mai 50200, Thailand
2
Office of Research Administration, Chiang Mai University, Chiang Mai 50200, Thailand
*
Authors to whom correspondence should be addressed.
Fermentation 2022, 8(4), 162; https://doi.org/10.3390/fermentation8040162
Submission received: 22 February 2022 / Revised: 31 March 2022 / Accepted: 1 April 2022 / Published: 3 April 2022
(This article belongs to the Special Issue Food Fermentation for Better Nutrition, Health and Sustainability)

Abstract

:
Fermented foods (FFs) hold global attention because of their huge advantages. Their health benefits, palatability, preserved, tasteful, and aromatic properties impart potential importance in the comprehensive evaluation of FFs. The bioactive components, such as minerals, vitamins, fatty acids, amino acids, and other phytochemicals synthesized during fermentation, provide consumers with several health benefits. Fermentation of food is an ancient process that has met with many remarkable changes owing to the development of scientific technologies over the years. Initially, fermentation relied on back-slapping. Nowadays, starter cultures strains are specifically chosen for the type of fermentation process. Modern biotechnological methods are being implemented in the fermentation process to achieve the desired product in high quality. Respiratory and gastrointestinal tract infections are the most severe health issues affecting human beings of all age groups, especially children and older adults, during this COVID-19 pandemic period. Studies suggest that the consumption of probiotic Lactobacillus strains containing fermented foods protects the subjects from common infectious diseases (CIDs, which is classified as upper respiratory tract infections, lower respiratory tract infections and gastrointestinal infections) by improving the host’s immune system. Further studies are obligatory to develop probiotic-based functional FFs that are effective against CIDs. Presently, we are urged to find alternative, safe, and cost-effective prevention measures against CIDs. The current manuscript briefs the production of FFs, functional properties of FFs, and their beneficial effects against respiratory tract infections. It summarizes the outcomes of clinical trials using human subjects on the effects of supplementation of FFs.

1. Introduction

Fermented foods state that “foods that have been transformed due to fermentation process via microbes such as bacteria, yeast, fungi and their enzymes”. The word fermentation is derived from the Latin word “fervere” meaning “to boil” [1] or “fermentare” meaning “to leaven” [2]. By simple means, fermented foods (FFs) are microbiologically processed raw materials (vegetables, meats, etc.). The International Scientific Association for Probiotics and Prebiotics defined FFs as “foods made through desired microbial growth and enzymatic conversions of food components” [3]. FFs are mainly available in the form of pickles, fermented juices, dairy, and meat products. The use of FFs varies depending on the availability of raw materials, region, and ethnicity [4]. FFs and beverages are enzymatically altered by the microorganisms in such a way that they taste and smell appealing to humans for consumption [5]. In addition, this improves the palatability of foods and insists on preservation [6]. The traditional preparation of FFs involves the natural fermentation process. The microorganisms responsible for the fermentation process define the products’ quality and type. Lactic acid bacteria (LAB), Bacillus spp., yeast, and molds are the commonly found critical microbes in fermented food products [7]. The FFs have been used to improve health status. Even though FFs have a long history, recently, due to increased attention from biologists, technologists, and consumers, many studies have been engaged in demonstrating that FFs can enhance gastrointestinal and systemic health [8,9,10,11]. The beneficial effects of the continuous consumption of FFs on inflammatory bowel disease [12], immunity boost [13], oxidative stress [14], cardiovascular diseases [15] cognitive enhancement [16], anxiety [17], and metabolic disorders [18] have been previously described. Nutritional intervention with fermented foods enhances immune function and alleviates upper respiratory tract infections (URTIs) [19], as well as improving intestinal and extra-intestinal health [20]. Zhang et al. [21] reported that daily consumption of Qingrun yogurt (fermented milk) for 12 weeks reduces the common cold in adults living in Northern China [21]. Ingestion of LAB-fermented food modulates the gut microbiome through their health promoting properties and production of metabolites such as biopeptides, vitamins, organic acids, bacteriocins, and anti-microbial compounds during fermentation [22].
Respiratory tract infections, such as URTIs and lower respiratory tract infections (LRTI), are a universal health threat among developed and developing countries [23]. LRTI, such as pneumonia, can be fatal in young and elderly individuals [24]. Viruses are responsible for causing more than 90% of URTIs [25]. The only way to prevent RTIs is to improve the immune function [19]. URTIs are infection in the mucosal surfaces of upper airways, such as the nose, sinuses, pharynx, or larynx, resulting in non-allergic rhinitis, acute sinusitis, acute pharyngitis/laryngitis, acute epiglottis, and acute otitis media [26,27,28]. Some of the viruses responsible for common respiratory infections are the influenza virus, respiratory syncytial virus, parainfluenza virus, rhinovirus, enterovirus, adenovirus, coronavirus, and others [28,29,30]. The influenza virus causes serious health threats, which may lead to morbidity [31]. The recent pandemic SARS-COV-2 virus and its associated respiratory infections create an alarming call about strengthening immune responses against a wide range of organisms that cause RTIs [32]. Acute respiratory infections in children is a serious health threat across the world, which also results in increased morbidity and mortality [33]. In this COVID-19 pandemic situation, respiratory and gastrointestinal tract infections are the most severe health issues affecting human beings of all age groups, especially children. Several treatments are available to treat respiratory infection [34]. However, a well-balanced diet, preferably comprised of probiotics or fermented functional foods, is also crucial for maintaining body homeostasis and fighting against respiratory and alimentary tract viruses. The probiotics in FFs enhance antiviral activity by producing bioactive compounds. These bioactive compounds improve immune function and lessen viral infections [35]. Naturally, the human body encompasses mechanical defense systems against the respiratory tract viruses, such as mucus, ciliated cells, and macrophages, which ward off the virus particles entering the system [36,37]. The LAB in FFs exhibit antiviral activity against the respiratory tract virus [38]. Certain FFs have proven to have anti-influenza functions. For example, kimchi, which contains Lactobacillus casei DK128, possesses antiviral activity against the influenza H3N2 virus [39]. The intervention of Lactobacillus rhamnosus GG in fermented milk product for 3 months demonstrated the prevention of respiratory discomforts in children attending daycare centers [40]. Probiotic delivery through fermentation is a combined approach; thus, probiotics in fermented foods produces added benefits and is advantageous over non-fermented products [41,42].
The current review summarizes the results of scientific reports on the beneficial effects of the consumption of FFs on respiratory infections and briefs the traditional and modern aspects of the production of FFs, functional properties of FFs, and their beneficial effects against respiratory tract infection. It summarizes the outcomes of clinical trials using human subjects on the effects of the supplementation of FFs. Peer-reviewed papers published in scientific journals were searched by using the keywords “Fermented foods”, “Respiratory infection”, “fermented food production”, “starter culture”, “probiotics in food”, and “health benefits of fermented foods” in PubMed, Scopus, Web of Science, and Google Scholar. The relevant papers were systemically screened and used to prepare the manuscript. Articles not published in the English language were not included in the study.

2. Traditional and Modern Aspects of the Production of FFs

Fermented food products, such as wine, beer, dairy products, and baked foods, are emerging globally and date back to 13,000 BC [43]. Fermentation can be categorized into indigenous fermentation (FFs produced traditionally by spontaneous fermentation) used for small scale production and technological fermentations (FFs produced using modern technological facilities) used for industrial scale production [2,43]. Extended shelf life, food safety, and improved organoleptic properties, such as aroma, taste, and texture, are important factors for fermentation practices [44]. The FFs play a prominent role in the socio-economic status of developing countries. The fermentation involves various biochemical processes by microorganisms and enzymes, resulting in significant food modification. The evolution of fermentation has been initiated ever since the beginning of human civilization. The traditional fermentation method is uncontrolled, as it depends on microorganisms from the environment [45]. The traditional fermentation process is of great economic value and reasonable food preservation method. Traditionally, the fermentation of foods occurs through two methods. Natural fermentation of foods and beverages are known as ‘wild ferments’ [46], where fermentation occurs by naturally available autochthonous or indigenous microbiota present in the food raw materials, which was subjected to changes depending on the surrounding environmental conditions, resulting in the characteristic FFs. The FFs are specific according to their geographic locations [43,47]. The second method is called ‘culture-dependent ferments’ [46,47,48,49,50,51]. Every new batch of fermentation was started with back-slopping, where a small amount of successful previous fermented batch product was used as an inoculum or natural starter culture [48].
The nutritional health benefits, increased food safety, and organoleptic features lead to the growing demand for FFs. Meanwhile, this leads to the increased awareness of food safety measures, standardization measures, industrial control of food production, starter cultures, and fermentation process control etiquettes at the industrial level [43]. The demand for FFs and increasing customers leads to an industrial standardization of fermentation. The first and prudent step is the selection of starter cultures, which is very important in determining the total fermentation process, their safety, organoleptic properties, and necessary changes in the food substrates [49]. From identifying various fermentation microbes and starter cultures, the progression of the development of fermentation techniques becomes pertinent. The current development in molecular biology techniques, next-generation sequencing (NGS), multi-omics and bioinformatics tools, and highly advanced statistical tools leads to further advancements, such as studying the genome sequence of industrially important microbes, their diversity, functions, and metabolic pathways. These outcomes result in great progress in the fermentation industry [50].
The term starter culture usually represents the prepared inoculum containing viable microorganisms that belongs to one or two or more than the two selected microbial species or strains. Thus, the desirable changes in the final product is obtained by using these selected commercial starter cultures while processing the raw materials to initiate the fermentation process. The purpose of starter cultures in fermentation is to fasten the process and to produce characteristic fermented foods. These starter cultures are specifically identified and isolated with the help of advanced microbiological techniques and are currently used in the industrial fermentation process to produce beverages such as beer, alcohol, and vinegar, and foods such as bread and other meat, as well as dairy products [52]. Such inoculation with selected microbes minimizes the risk of foodborne diseases [53,54]. The starter cultures had also been selected from the screened isolates, which yielded end products with high organoleptic features in previous fermentation processes [55]. Molecular approaches, such as high-throughput screening (HTS) of target genes, genetic engineering of specific and well-adapted starter cultures, are synthesized and incorporated to create better-improved fermentation [52].
The starter cultures are improved through another novel technology, CRISPR/Cas9 (Clustered Regularly Interspaced Short Palindromic Repeats/CRISPR-associated protein 9), where the genome of the specific target microbe is precisely edited [56]. Thus, the microbiome manipulation using CRISPR-based technologies improves specific genes in starter cultures [57]. Advanced genetic engineering can rule out undesirable microbial populations and enhance the desirable microbiota, leading to optimized FFs [58]. Recent scientific advances could aid in developing the whole food microbial ecosystem, their interactions, and pathways owing to desirable fermentation processes [59]. Despite the desired quality of starter cultures produced through recombinant DNA technology, those strains, and their genetically modified food ingredients are restricted from usage due to stern food regulations and a lack of acceptance from consumers [60]. Various other technologies include dominant selection, random mutagenesis, natural competence, and conjugation [61].
In the traditional method, the microbial composition of the fermented products depends on the microbial culture used. Outdated plate cultivation method is unable to provide accurate microbial profile information; henceforth, recent NGS technology encompassing collective studies of metagenomics, meta-transcriptomics, meta-proteomics, and metabolomics enables the identification of accurate microbiome from different microbial communities, which pave the way to the detailed study of microbe–microbe interactions in fermentation ecosystems [62]. FFs contain different types of microbial ecosystems such as bacteria, fungi, and yeast; altogether, they are responsible for the quality and safety of FFs [63]. Integrated multi-omics technologies, next-generation nucleic acid, and protein studies enhance the microbes used for the fermentation process, and provide improved fermented products with prolonged shelf life and desirable characteristics. The stability and safety of fermentation and FFs have been improved. The genome editing and molecular approaches rapidly improved the microbial functions, resulting in better quality products with increased safety and an extended shelf life. The next-generation nucleic acid and protein-based approaches provide adequate details about the microbial community for better fermentation and desired products with more shelf life [64].

3. Functional Properties of Fermented Food and Beneficial Effects against Respiratory Tract Infection

By definition, FFs can be either plant or animal-based and are produced through back-slopping or by spontaneous fermentation via enzymatic actions of microorganisms so that raw materials are converted into FFs. FFs are rich in nutrients with better taste and provide health benefits [65,66]. Every so often, FFs and beverages are supplemented with probiotics to improve nutritional and health properties [67]. FFs are familiar because of their health benefits and safety standards. Not all FFs have live organisms; in some products, they have been heat-treated, inactivating the microbes. Moreover, FFs are a rich source of bioactive microbes [14]. FFs are functional foods that generally boost the immune functions and metabolism of the consumer [51]. Probiotics are used to ferment the foods traditionally, hence, probiotics and fermented foods are closely related [68]. During fermentation, various bioactive peptides, oligosaccharides, lipids, and other components are synthesized. These components improve the functionality of the product, such as antioxidants, antihypertensive, and other bioactivities [69,70,71]. The health benefits of FFs include preventing metabolic diseases, cardiovascular diseases, enhancing immune function, and improving cognition [12,14,15,17,72]. In 1907, Metchnikoff explained the health benefits of fermented milk products [73]. The traditional fermented milk products are highly rich in bioactive compounds such as peptides, amino acids, vitamins, and minerals [74]. Bioactive compounds are found in unprocessed counterparts such as organic acids, fatty acids, bacteriocins, amino acids, and exopolysaccharides [75,76]. Naturally, bioactive compounds are being released into the system in two different ways. One is that it is released from raw materials because of high acidity. Another way is due to the hydrolyzing enzymes of the probiotic microflora [77]. The FFs are involved in the improvement of intestinal permeability and barrier function [78] and demonstrate positive effects on atherosclerosis, inflammatory bowel diseases, colon cancer [79], anger, depression, and anxiety [80]. FFs aid in maintaining the gut microflora with the presence of bioactive chemicals, neuropeptides, antioxidants, and anti-inflammatory activity [81].
Probiotic bacteria bind directly to the virus and disrupt the adhesion of virus cells onto the mucosal cells [82]. Muhialdin et al. reviewed the connection between FFs, their probiotics with the anti-viral mechanisms and the immune system, and proficient antiviral activity against respiratory and alimentary tract ailments [35]. The antiviral mechanisms involve various systemic changes such as enhancing natural killer cells (NK cells), pro-inflammatory cytokines, and cytotoxic T lymphocytes (CD3+, CD16+, and CD56+) [83]. Bioactive yogurt compounds resist upper respiratory tract infections [84,85,86,87,88] and remain potentially beneficial by preventing the common cold and influenza [89,90].
Gouda et al. [91] reviewed a few studies [92,93,94,95] and stated that yogurt bioactive peptides and probiotics exhibited antiviral properties against respiratory infectious viruses that share some mechanistic similarities with the SARS-CoV-2 virus [91]. Cell-free supernatants of the metabolites of yogurt fermented with Lactobacillus plantarum and yogurt fermented with Bifidobacterium bifidum demonstrated anti-enterovirus 71 activity [92]. Metabolites of both L. casei, and Bifidobacterium adolescentis demonstrated effective antiviral activity against a rotavirus infection [93]. In vitro studies by Starosila et al. [94] described that the influenza virus was inhibited completely by the P18 peptide produced by Bacillus subtilis. Similarly, another research study [95] in mice stated that Lactobacillus gasseri SBT2055 prevents the influenza A virus and respiratory syncytial viral infections [95]. As per the review provided by Gouda et al. [91], the yogurt bioactive peptides are effective against the COVID-19 virus through the inhibition of the angiotensin-converting enzyme (ACE) and might influence the COVID-19 presentation and outcome [91].
Korean traditionally fermented foods that contain grains, fruits, herbs, and mushrooms provide high levels of Lactobacilli that undergo lactic acid fermentation and produce different metabolites that exhibit beneficial health effects [96] such as crucial antiviral properties against the influenza virus [97]. Probiotic LAB or the fermented foods containing probiotic LAB elicit an antagonist effect on the influenza virus by stimulating immune responses by increasing interferon (IFN-α, IFN-β, and IFN-γ) and interleukin (IL-2, IL-6, and IL-12) levels [98]. L. plantarum (YML009) [99] and L. plantarum (DK119) [97] isolated from kimchi were found to have antiviral activity against the H1N1 influenza virus [97,99]. The yogurt containing L. lactis reduces influenza symptoms [100]. Japanese-fermented foods containing L. plantarum YU mitigate influenza virus replication [101]. L. plantarum DK119 from kimchi was proven to demonstrate complete protection against influenza-A viruses [97]. Oral administration of fermented yoghurt containing Lactobacillus bulgaricus OLL1073R-1 or its exopolysaccharides ameliorates the influenza viral infection by developing splenocytic NK cell activity [102]. L. plantarum and Leuconosoc mesenteroides demonstrated antiviral activity against the H1N1 and H1N9 influenza virus [103].
The supplementation of 65 mL of probiotic fermented milk containing L. rhamnosus GG (ATCC 53103) (7.1 × 109 CFU per day), Bifidobacterium sp. B420 (8.4 × 109 CFU per day), Lactobacillus acidophilus 145 (3.2 × 109 CFU per day), and Streptococcus thermophilus (27 × 109 CFU per day) to human volunteers (patients with nasal potentially pathogenic bacteria; n = 108; 33 females, 75 males; age = 41 ± 8 years old) for 28 days reduced the colonization of potent nasal pathogenic bacteria (Staphylococcus aureus, Streptococcus pneumoniae, and β-hemolytic streptococci) compared to that of the control group (patients with nasal potentially pathogenic bacteria; n = 101; 29 females, 72 males; age = 39 ± 9 years old) supplemented with standard yogurt (180 g containing conventional LAB strains such as Lactobacillus delbrueckii subsp. bulgaricus and S. thermophilus; total ≥ 107 CFU per gram). The study proposed that the oral consumption of probiotic fermented milk may stimulate the B lymphocytes of gut-associated lymphoid tissue, which may further reach the upper respiratory system and elucidate secretory IgA production [104].
Shift-based workers (n = 500; 43% males; age = 31.8 ± 8.9 years old) [84] and free-living elderly (n = 537; 339 females, 198 males; age range = 72–80 years old) [105] were supplemented with a fermented dairy product (Actimel®; dosage 200 g per day) containing the probiotic L. casei DN-114001 (≥ 1010 CFU/100 g of product) and the common starter culture L. delbrueckii subsp. bulgaricus and S. thermophilus (total CFU of starter cultures ≥ 109 CFU per 100 g) for three months, and the effects on common respiratory and gastrointestinal infectious diseases (CIDs) were monitored [84,105]. The total study period includes 2 weeks of diet control prior to probiotic supplementation, 3 months of probiotic supplementation and a 1-month follow-up period without probiotic supplementation [84,105]. The probiotic supplementation reduced the incidence of CIDs, cumulated duration of fever, cumulated number of CIDs (in the subgroup of smokers), and increased the time to the first incidence of CIDs in the probiotic group. The probiotic supplementation improved the immune cell (leukocyte, neutrophil, and natural killer) counts and its activity in shift-based workers compared to that of the control group (n = 500; 44% males; age = 32.5 ± 8.9 years old). The study indicated that the daily consumption of Actimel® could reduce the incidence and severity of CIDs in stressed individuals [84]. Similarly, the supplementation of Actimel® significantly reduced the average period per episode and cumulative duration of CIDs in elderly subjects compared to the placebo control group (n = 535; 333 females, 202 males; age range = 73–81 years old) supplemented with non-fermented dairy product. The probiotic-supplemented group significantly reduced both the cumulative durations and episodes of URTIs (upper respiratory tract infections) and rhinopharyngitis. The fecal load of L. casei was also found to be high in the probiotic group. The study proved that the fermented product was safe, and regular consumption of fermented products made with the probiotic L. casei DN-114 001 could reduce the incidence of CIDs, especially URTIs, in elderly subjects [105]. Healthy children (n = 314; 157 females, 157 males; age range = 3 to 6 years old) attending daycare centers/schools were supplemented with Actimel® for 3 months, and the incidence of CIDs and behavioral changes due to illness were assessed. The intervention with Actimel® for three months effectively reduced the incidence of CIDs in children with no differences in behavioral changes due to illness compared to that of the control group (n = 324; 152 females, 172 males; age range = 3 to 6 years old) supplemented with the non-fermented acidified dairy product [106].
Healthy children (n = 139; 61 females, 78 males; age range = 13 to 86 months old) attending daycare centers were supplemented with 100 mL of fermented milk product containing 109 CFU of the probiotic L. rhamnosus strain GG (LGG) for three months. The study demonstrated that the LGG supplementation significantly reduced the risk, duration, and symptoms of URTIs compared to that of the placebo group (n = 142; 63 females, 79 males; age range = 13 to 83 months old) supplemented with post-pasteurized fermented milk product without LGG. No changes were observed in the incidence and duration of gastrointestinal infections, vomiting, and diarrheal episodes among the study subjects. Thus, daily intake of LGG reduces the URTIs in children attending daycare centers [40].
Fujita et al. [85] investigated the effect of daily consumption of fermented milk (dosage 80 mL per day) containing the probiotic L. casei strain Shirota (4 × 1010 CFU/80 mL of product) on URTIs in elderly (n = 76; 57 females, 19 males; age = 83.5 ± 8.9 years old) at daycare facilities. The daily intake of probiotic reduced the duration of infection in elderly compared to that of the placebo group (n = 78; 52 females, 26 males; age = 83.0 ± 9.3 years old) supplemented with the placebo drink (same drink without probiotic L. casei strain Shirota) [85].
Makino et al. [89] evaluated the effect of the intake of yoghurt (dosage: 90 g per day; for 8 weeks at the Funagata study or for 12 weeks at the Arita study) fermented with the probiotic L. delbrueckii subsp. bulgaricus OLL1073R-1 (2.0–3.5 × 108 CFU per gram) and S. thermophilus OLS3059 (6.3–8.8 × 108 CFU per gram) on the common cold in healthy elderly subjects. In the Funagata study, 57 elderlies were selected (age range = 69–80 years old) and assigned for the probiotic group (n = 29) and milk group (n = 28). In the Arita study, 101 elderlies were selected (age range = 59–84 years old) and assigned for the probiotic group (n = 43) and milk group (n = 42). Both studies in the probiotic group exhibited reduction in the risk of catching the common cold compared to that of the milk group supplemented with milk (dosage 100 mL per day for 8 or 12 weeks). The probiotic intake also increased the quality of life score for the eye/nose/throat system, which was correlated with the improved natural killer cell activity in elderly compared to that of the milk group [89].
Healthy children (1–4 years old) attending daycare or preschool were supplemented with fermented cow’s milk (FCM) containing Lactobacillus paracasei CBA L74 (5.9 × 109 CFU/g) in powder form (the live microbes are heat-killed before spray drying the fermented milk product) (daily dosage: 7 g of powder diluted in 150 mL of cow’s milk or water) for three months, and the incidence of CIDs and changes in immunity was assessed. The FCM supplementation significantly reduced the incidence of CIDs and URTIs and decreased acute gastroenteritis in children of the probiotic group (n = 66) compared to that of the placebo group (n = 60) supplemented with maltodextrins. Changes in the innate and acquired immune system were also recorded in FCM-treated groups. The study concluded that FCM containing the heat-killed CBA L74 could improve their immunity in children [107]. Another study by Nocerino et al. also reported that the supplementation of FCM and fermented rice product containing L. paracasei CBA L74 (5.9 × 109 CFU per gram; heat-killed) prevents CIDs’ incidence by prompting the innate and acquired immune system in children (age = 23 ± 10 months old; FCM group n = 137; fermented rice product group n = 118) compared to that of the placebo group (n = 122) supplemented with maltodextrins [108].
A twelve-week intervention with fermented milk containing L. casei strain Shirota (1.0 × 1011 CFU per day) significantly reduced the incidence, duration, and symptoms of URTIs in middle-aged (30–49 years old) office workers of the probiotic group (n = 49) compared to that of the control group (n = 47) supplemented with milk. The study also revealed that the probiotic supplementation activated the NK cells. Both the control and probiotic group exhibited increased salivary cortisol secretion. The results suggest that the daily consumption of fermented milk containing L. casei strain Shirota may reduce the risk of URTIs in middle-aged office workers [109]. Mai et al. reported that consuming fermented milk containing L. casei strain Shirota (1.0 × 108 CFU per ml; dosage = 65 mL per day) for 12 weeks reduces the incidence of diarrhea, constipation, and acute respiratory tract infection in children (n = 510; 236 females, 274 males; age range = 3–5 years old) compared to that of the control group (n = 493; 209 females, 284 males; age range = 3–5 years old) [110] (Table 1).

4. Conclusions

The daily consumption of probiotic-based fermented products could improve the host’s health status and aid in resisting respiratory infections. Advanced and powerful biotechnological tools are used to calibrate and standardize fermentation processes and provide more desirable fermented foods with enhanced quality, taste, aroma, long shelf life, nutritional content, and safety, and prevent contamination by spoilage organisms. Fermentation has undergone remarkable changes over the years. Genome editing of the microbiome through NGS and CRISPR/Cas9 techniques are efficiently employed in fermentation industries to improve the functionality of the microbial community in different cultures. In addition to the wide range of applications of fermentation in the food industry, this technology was also found to be prominent in treating some diseases such as immune, respiratory, gastrointestinal diseases, and common infectious diseases. A minimal number of clinical trials have been conducted using human subjects on the effects of fermented food supplementation on CIDs.
FFs used in all the clinical trials that are included in this review resulted to be probiotics supplemented with fermented dairy products that exhibited beneficial effects against respiratory tract infections in children, adults, and elderly individuals. Among the reviewed studies reporting the probiotics used in the intervention of FFs, L. casei [84,85,105,106,109,110] and L. paracasei (heat-killed) [107,108] are the most commonly used probiotics for health-promoting properties. The results supported the assertion that regular consumption of probiotic-containing fermented drinks could reduce the risk of CIDs [84,105,106,107,108,110] and URTIs [40,84,85,89,104,105,107,108,109,110], especially in children [40,106,107,108,110]. The possible mechanisms behind the protective effect of probiotic-based fermented products are the stimulation of the host’s innate and acquired immune system. The studies suggested that the consumption of probiotic Lactobacillus strains containing fermented foods protected the subjects from CIDs and upper respiratory tract infections by improving the host’s immune system. However, many clinical trial studies with fermented food products with antiviral properties are further required to know the complete mechanisms of the probiotic FF products for its use, in order to develop conventional probiotic-based functional foods that are effective against respiratory tract infections.

Author Contributions

Conceptualization, B.S.S., P.K. and C.C.; methodology, P.K. and S.T.; validation, B.S.S., S.T., P.K. and C.C.; formal analysis, S.T. and B.S.S.; investigation, S.T. and B.S.S.; resources, C.C.; writing—original draft preparation, S.T., B.S.S., P.K. and C.C.; writing—review and editing, S.T., B.S.S., P.K., M.B. and C.C.; supervision, C.C. and B.S.S.; project administration, C.C.; funding acquisition, C.C. All authors have read and agreed to the published version of the manuscript.

Funding

This research received funding from the Chiang Mai University, Thailand.

Institutional Review Board Statement

Not applicable.

Informed Consent Statement

Not applicable.

Data Availability Statement

Not applicable.

Acknowledgments

We wish to thank the Faculty of Pharmacy, Chiang Mai University, Chiang Mai, Thailand. The study was partially supported by the Chiang Mai University, Chiang Mai, Thailand.

Conflicts of Interest

The authors declare no conflict of interest.

References

  1. Fruton, J. Fermentation: Vital or chemical process? In History of Science and Medicine Library; Brill: Leiden-Boston, The Netherlands, 2006; Volume 1, p. 141. ISBN 978-90-04-15268-7. [Google Scholar]
  2. Martínez-Espinosa, R.M. Introductory Chapter: A Brief Overview on Fermentation and Challenges for the Next Future. In New Advances on Fermentation Processes; IntechOpen: London, UK, 2020. [Google Scholar] [CrossRef] [Green Version]
  3. Marco, M.L.; Sanders, M.E.; Gänzle, M.; Arrieta, M.C.; Cotter, P.D.; De Vuyst, L.; Hill, C.; Holzapfel, W.; Lebeer, S.; Merenstein, D.; et al. The International Scientific Association for Probiotics and Prebiotics (ISAPP) consensus statement on fermented foods. Nat. Rev. Gastroenterol. Hepatol. 2021, 18, 196–208. [Google Scholar] [CrossRef] [PubMed]
  4. Sivamaruthi, B.S.; Kesika, P.; Prasanth, M.I.; Chaiyasut, C. A mini review on antidiabetic properties of fermented foods. Nutrients 2018, 10, 1973. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  5. Steinkraus, K.H. Classification of fermented foods: Worldwide review of household fermentation techniques. Food Control. 1997, 8, 311–317. [Google Scholar] [CrossRef]
  6. Azokpota, P.S. Quality aspects of alkaline-fermented foods. In Handbook of Indigenous Foods Involving Alkaline Fermentation; Sarkar, P.K., Nout, M.J.R., Eds.; CRC Press Taylor & Francis Group: Boca Raton, FL, USA, 2015; pp. 315–379. [Google Scholar]
  7. Marco, M.L.; Heeney, D.; Binda, S.; Cifelli, C.J.; Cotter, P.D.; Foligné, B.; Gänzle, M.; Kort, R.; Pasin, G.; Pihlanto, A.; et al. Health benefits of fermented foods: Microbiota and beyond. Curr. Opin. Biotechnol. 2017, 44, 94–102. [Google Scholar] [CrossRef] [PubMed]
  8. Rosa, D.D.; Dias, M.M.S.; Grześkowiak, Ł.M.; Reis, S.A.; Conceição, L.L.; Peluzio, M.D.C.G. Milk kefir: Nutritional, microbiological and health benefits. Nutr. Res. Rev. 2017, 30, 82–96. [Google Scholar] [CrossRef] [PubMed]
  9. Şanlier, N.; Gökcen, B.B.; Sezgin, A.C. Health benefits of fermented foods. Crit. Rev. Food Sci. Nutr. 2019, 59, 506–527. [Google Scholar] [CrossRef]
  10. Lavefve, L.; Marasini, D.; Carbonero, F. Microbial Ecology of Fermented Vegetables and Non-Alcoholic Drinks and Current Knowledge on Their Impact on Human Health. Adv. Food Nutr. Res. 2019, 87, 147–185. [Google Scholar] [PubMed]
  11. Mathur, H.; Beresford, T.P.; Cotter, P.D. Health benefits of lactic acid bacteria (LAB) fermentates. Nutrients 2020, 12, 1679. [Google Scholar] [CrossRef]
  12. Lorea Baroja, M.; Kirjavainen, P.V.; Hekmat, S.; Reid, G. Anti-inflammatory effects of probiotic yogurt in inflammatory bowel disease patients. Clin. Exp. Immunol. 2007, 149, 470–479. [Google Scholar] [CrossRef]
  13. Ashaolu, T.J. Immune boosting functional foods and their mechanisms: A critical evaluation of probiotics and prebiotics. Biomed. Pharmacother. 2020, 130, 110625. [Google Scholar] [CrossRef]
  14. Sivamaruthi, B.S.; Kesika, P.; Chaiyasut, C. A comprehensive review on functional properties of fermented rice bran. Pharmacogn. Rev. 2018, 12, 218–224. [Google Scholar] [CrossRef]
  15. Tapsell, L.C. Fermented dairy food and CVD risk. Br. J. Nutr. 2015, 113 (Suppl. 2), S131–S135. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  16. Sivamaruthi, B.S.; Kesika, P.; Chaiyasut, C. Impact of fermented foods on human cognitive function-a review of outcome of clinical trials. Sci. Pharm. 2018, 86, 22. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  17. Hilimire, M.R.; De Vylder, J.E.; Forestell, C.A. Fermented foods, neuroticism, and social anxiety: An interaction model. Psychiatry Res. 2015, 228, 203–208. [Google Scholar] [CrossRef] [PubMed]
  18. Iwasa, M.; Aoi, W.; Mune, K.; Yamauchi, H.; Furuta, K.; Sasaki, S.; Takeda, K.; Harada, K.; Wada, S.; Nakamura, Y.; et al. Fermented milk improves glucose metabolism in exercise-induced muscle damage in young healthy men. Nutrition 2013, 12, 83. [Google Scholar] [CrossRef] [Green Version]
  19. Jespersen, L.; Tarnow, I.; Eskesen, D.; Morberg, C.M.; Michelsen, B.; Bugel, S.; Dragsted, L.O.; Rijkers, G.T.; Calder, P.C. Effect of Lactobacillus paracasei subsp. paracasei, L. casei 431 on immune response to influenza vaccination and upper respiratory tract infections in healthy adult volunteers: A randomized, double-blind, placebo-controlled, parallel-group study. Am. J. Clin. Nutr. 2015, 101, 1188–1196. [Google Scholar]
  20. Kok, C.R.; Hutkins, R. Yogurt and other fermented foods as sources of health-promoting bacteria. Nutr. Rev. 2018, 76, 4–15. [Google Scholar] [CrossRef] [Green Version]
  21. Zhang, H.; Miao, J.; Su, M.; Liu, B.Y.; Liu, Z. Effect of fermented milk on upper respiratory tract infection in adults who lived in the haze area of Northern China: A randomized clinical trial. Pharm. Biol. 2021, 59, 647–652. [Google Scholar] [CrossRef]
  22. Castellone, V.; Bancalari, E.; Rubert, J.; Gatti, M.; Neviani, E.; Bottari, B. Eating Fermented: Health Benefits of LAB-Fermented Foods. Foods 2021, 31, 2639. [Google Scholar] [CrossRef]
  23. WHO Organization. World Health Report 2004 Statistical Annex; WHO: Geneva, Switzerland, 2004. [Google Scholar]
  24. Jain, S. Epidemiology of viral pneumonia. Clin. Chest Med. 2017, 38, 1–9. [Google Scholar] [CrossRef]
  25. Baud, D.; Agri, V.D.; Gibson, G.R.; Reid, G.; Giannoni, E. Using probiotics to flatten the curve of coronavirus disease COVID-2019 pandemic. Front. Public Health 2020, 8, 186. [Google Scholar] [CrossRef] [PubMed]
  26. Hao, Q.; Lu, Z.; Dong, B.R.; Huang, C.Q.; Wu, T. Probiotics for preventing acute upper respiratory tract infections. Cochrane Database Syst. Rev. 2011, 7, CD006895. [Google Scholar]
  27. Dasaraju, P.V.; Liu, C. Infections of the Respiratory System, In Medical Microbiology, 4th ed.; Baron, S., Ed.; University of Texas Medical Branch at Galveston: Galveston, TX, USA, 1996; p. 65. [Google Scholar]
  28. Meneghetti, A. Upper Respiratory Tract Infection Medication. Medscape 2015, 11, 1–18. [Google Scholar]
  29. Boncristiani, H.F.; Criado, M.F.; Arruda, E. Respiratory Viruses. In Encyclopedia of Microbiology, 3rd ed.; Schaechter, M., Ed.; Elsevier Inc: Amsterdam, The Netherlands, 2009; pp. 500–518. [Google Scholar]
  30. Kompanikova, J.; Zumdick, A.; Neuschlova, M.; Sadlonova, V.; Novakova, E. Microbiologic methods in the diagnostics of upper respiratory tract pathogens. Clin. Res. Pract. 2017, 1020, 25–31. [Google Scholar]
  31. Taubenberger, J.K.; Morens, D.M. The pathology of influenza virus infections. Annu. Rev. Pathol. 2008, 3, 499–522. [Google Scholar] [CrossRef] [PubMed]
  32. BourBour, F.; Mirzaei Dahka, S.; Gholamalizadeh, M.; Akbari, M.E.; Shadnoush, M.; Haghighi, M.; Masoumi, H.T.; Ashoori, N.; Doaei, S. Nutrients in prevention, treatment, and management of viral infections; special focus on Coronavirus. Arch. Physiol. Biochem. 2020, 1–10. [Google Scholar] [CrossRef]
  33. Mulholland, K. Global burden of acute respiratory infections in children: Implications for interventions. Pediatr. Pulmonol. 2003, 36, 469–474. [Google Scholar] [CrossRef]
  34. Abed, Y.; Boivin, G. Treatment of respiratory virus infections. Antivir. Res. 2006, 70, 1–16. [Google Scholar] [CrossRef]
  35. Muhialdin, B.J.; Zawawi, N.; Abdull Razis, A.F.; Bakar, J.; Zarei, M. Antiviral activity of fermented foods and their probiotics bacteria towards respiratory and alimentary tracts viruses. Food Control 2021, 127, 108140. [Google Scholar] [CrossRef]
  36. Gizurarson, S. The effect of cilia and the mucociliary clearance on successful drug delivery. Biol. Pharm. Bull. 2015, 38, 497–506. [Google Scholar] [CrossRef] [Green Version]
  37. He, W.; Chen, C.J.; Mullarkey, C.E.; Hamilton, J.R.; Wong, C.K.; Leon, P.E.; Uccellini, M.B.; Chromikova, V.; Henry, C.; Hoffman, K.W.; et al. Alveolar macrophages are critical for broadly-reactive antibody-mediated protection against influenza A virus in mice. Nat. commun. 2017, 8, 846. [Google Scholar] [CrossRef] [PubMed]
  38. Seo, D.J.; Day, J.; Jung, S.; Yeo, D.; Choi, C. Inhibitory effect of lactic acid bacteria isolated from kimchi against murine norovirus. Food Control. 2019, 109, 106881. [Google Scholar] [CrossRef]
  39. Jung, Y.J.; Lee, Y.T.; Ngo, V.L.; Cho, Y.H.; Ko, E.J.; Hong, S.M.; Kim, K.H.; Jang, J.H.; Oh, J.S.; Park, M.K.; et al. Heat-killed Lactobacillus casei confers broad protection against influenza A virus primary infection and develops heterosubtypic immunity against future secondary infection. Sci. Rep. 2017, 7, 17360. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  40. Hojsak, I.; Snovak, N.; Abdović, S.; Szajewska, H.; Misak, Z.; Kolacek, S. Lactobacillus GG in the prevention of gastrointestinal and respiratory tract infections in children who attend day care centers: A randomized, double-blind, placebo-controlled trial. Clin. Nutr. 2010, 29, 312–316. [Google Scholar] [CrossRef] [PubMed]
  41. Ranadheera, C.; Vidanarachchi, J.; Rocha, R.; Cruz, A.; Ajlouni, S. Probiotic Delivery through Fermentation: Dairy vs. Non-Dairy Beverages. Fermentation 2017, 3, 67. [Google Scholar] [CrossRef] [Green Version]
  42. Baliyan, N.; Kumari, M.; Kumari, P.; Dindhoria, K.; Mukhia, S.; Kumar, S.; Gupta, M.; Kumar, R. Probiotics in fermented products and supplements. In Current Developments in Biotechnology and Bioengineering; Rai, A.K., Sudhir, S.P., Pandey, A., Larroche, C., Soccol, C.R., Eds.; Elsevier: Amsterdam, The Netherlands, 2022; pp. 73–107. [Google Scholar]
  43. Galimberti, A.; Bruno, A.; Agostinetto, G.; Casiraghi, M.; Guzzetti, L.; Labra, M. Fermented food products in the era of globalization: Tradition meets biotechnology innovations. Curr. Opin. Bioethanol. 2021, 70, 36–41. [Google Scholar] [CrossRef]
  44. Chojnacka, K. Fermentation products. Chem. Eng. Chem. Process Technol. 2010, 5, 189–200. [Google Scholar]
  45. Adesulu, A.T.; Awojobi, K.O. Enhancing sustainable development through indigenous fermented food products in Nigeria. Afr. J. Microbiol. Res. 2014, 8, 1338–1343. [Google Scholar]
  46. Dimidi, E.; Cox, R.S.; Rossi, M.; Whelan, K. Fermented foods: Definitions and characteristics, impact on the gut microbiota and effects on gastrointestinal health and disease. Nutrients 2019, 11, 1806. [Google Scholar] [CrossRef] [Green Version]
  47. Giraffa, G. Studying the dynamics of microbial populations during food fermentation. FEMS Microbiol. Rev. 2004, 28, 251–260. [Google Scholar] [CrossRef] [Green Version]
  48. Whittington, H.D.; Dagher, S.F.; Bruno-Bárcena, J.M. Production and Conservation of Starter Cultures: From “Backslopping” to Controlled Fermentations. In How Fermented Foods Feed a Healthy Gut Microbiota; Azcarate-Peril, M., Arnold, R., Bruno-Bárcena, J., Eds.; Springer: Cham, Switzerland, 2019; pp. 125–138. [Google Scholar]
  49. Gilliland, S.E. Bacterial Starter Cultures for Food, 1st ed.; CRC Press: Boca Raton, FL, USA, 2018. [Google Scholar]
  50. Ojha, K.S.; Tiwari, B.K. Novel food fermentation technologies. In Novel Food Fermentation Technologies; Springer: Cham, Switzerland, 2016; pp. 71–87. [Google Scholar]
  51. Rezac, S.; ReenKok, C.; Heermann, M.; Hutkins, R.W. Fermented foods as a dietary source of live organisms. Front. Microbiol. 2018, 24, 1785. [Google Scholar] [CrossRef] [PubMed]
  52. Hansen, E.B. Commercial bacterial starter cultures for fermented foods of the future. Int. J. Food Microbiol. 2002, 78, 119–131. [Google Scholar] [CrossRef]
  53. Gadaga, T.H.; Nyanga, L.K.; Mutukumira, A.N. The occurrence, growth and control of pathogens in African fermented foods. Afr. J. Food Agric. Nutr. Dev. 2004, 4, 5346. [Google Scholar] [CrossRef]
  54. Panagou, E.Z.; Tassou, C.C.; Vamvakoula, P.; Saravanos, E.K.; Nychas, G.J.E. Survival of Bacillus cereus vegetative cells during Spanish-style fermentation of conservolea green olives. J. Food Prot. 2008, 71, 1393–1400. [Google Scholar] [CrossRef] [PubMed]
  55. Vinicius De Melo Pereira, G.; De Carvalho Neto, D.P.; Junqueira, A.C.D.O.; Karp, S.G.; Letti, L.A.; Magalhães Júnior, A.I.; Soccol, C.R. A review of selection criteria for starter culture development in the food fermentation industry. Food Rev. Int. 2020, 36, 135–167. [Google Scholar] [CrossRef]
  56. Ran, F.A.; Hsu, P.D.; Wright, J.; Agarwala, V.; Scott, D.A.; Zhang, F. Genome engineering using the CRISPR-Cas9 system. Nat. Protoc. 2013, 8, 2281–2308. [Google Scholar] [CrossRef] [Green Version]
  57. Ramachandran, G.; Bikard, D. Editing the microbiome the CRISPR way. Philos. Trans. R. Soc. 2019, 374, 20180103. [Google Scholar] [CrossRef] [Green Version]
  58. Barrangou, R.; Notebaart, R.A. CRISPR-directed microbiome manipulation across the food supply chain. Trends Microbiol. 2019, 27, 489–496. [Google Scholar] [CrossRef]
  59. Mays, Z.J.; Nair, N.U. Synthetic biology in probiotic lactic acid bacteria: At the frontier of living therapeutics. Curr. Opin. Biotechnol. 2018, 53, 224–231. [Google Scholar] [CrossRef]
  60. Johansen, E. Future access and improvement of industrial lactic acid bacteria cultures. Microb. Cell Factories 2017, 16, 230. [Google Scholar] [CrossRef] [Green Version]
  61. Derkx, P.M.; Janzen, T.; Sørensen, K.I.; Christensen, J.E.; Stuer-Lauridsen, B.; Johansen, E. The art of strain improvement of industrial lactic acid bacteria without the use of recombinant DNA technology. Microb. Cell Factories 2014, 13, S5. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  62. Zhao, M.; Su, X.Q.; Nian, B.; Chen, L.J.; Zhang, D.L.; Duan, S.M.; Wang, L.Y.; Shi, X.Y.; Jiang, B.; Jiang, W.W.; et al. Integrated meta-omics approaches to understand the microbiome of spontaneous fermentation of traditional Chinese pu-erh tea. Msystems 2019, 4, e00680–e00719. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  63. Wolfe, B.E.; Dutton, R.J. Fermented foods as experimentally tractable microbial ecosystems. Cell 2015, 161, 49–55. [Google Scholar] [CrossRef] [Green Version]
  64. Ferrocino, I.; Cocolin, L. Current perspectives in food-based studies exploiting multi-omics approaches. Curr. Opin. Food Sci. 2017, 13, 10–15. [Google Scholar] [CrossRef] [Green Version]
  65. Ghosh, T.; Beniwal, A.; Semwal, A.; Navani, N.K. Mechanistic Insights into Probiotic Properties of Lactic Acid Bacteria Associated with Ethnic Fermented Dairy Products. Front. Microbiol. 2019, 10, 502. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  66. Hutkins, R.W. Microbiology and Technology of Fermented Foods, 2nd ed.; Hutkins, R.W., Ed.; Wiley-Blackwell: Hoboken, NJ, USA, 2019; p. 624. [Google Scholar]
  67. Gille, D.; Schmid, A.; Walther, B.; Vergères, G. Fermented Food and Non-Communicable Chronic Diseases: A Review. Nutrients 2018, 10, 448. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  68. Mahasneh, A.M.; Abbas, M.M. Probiotics and traditional fermented foods: The eternal connection (mini-review). Jordan J. Biol. Sci. 2010, 3, 133–140. [Google Scholar]
  69. Hayes, M.; García-Vaquero, M. Bioactive compounds from fermented food products. In Novel Food Fermentation Technologies, 1st ed.; Shikha Ojha, K., Tiwari, B.K., Eds.; Springer: Cham, Switzerland, 2016; pp. 293–310. [Google Scholar]
  70. Park, Y.W.; Nam, M.S. Bioactive Peptides in Milk and Dairy Products: A Review. Korean J. Food Sci. Anim. Resour. 2015, 35, 831–840. [Google Scholar] [CrossRef] [Green Version]
  71. Gianfranceschi, G.L.; Gianfranceschi, G.; Quassinti, L.; Bramucci, M. Biochemical requirements of bioactive peptides for nutraceutical efficacy. J. Funct. Foods 2018, 47, 252–263. [Google Scholar] [CrossRef]
  72. An, S.Y.; Lee, M.S.; Jeon, J.Y.; Ha, E.S.; Kim, T.H.; Yoon, J.Y.; Ok, C.O.; Lee, H.K.; Hwang, W.S.; Choe, S.J.; et al. Beneficial effects of fresh and fermented kimchi in prediabetic individuals. Ann. Nutr. Metab. 2013, 63, 111–119. [Google Scholar] [CrossRef]
  73. Anukam, K.C.; Reid, G. Probiotics: 100 years (1907–2007) after Elie Metchnikoff’s observation. In Communicating Current Research and Educational Topics and Trends in Applied Microbiology, 1st ed.; Mendez Vilas, A., Ed.; Formatex: Badajoz, Spain, 2007; pp. 466–474. [Google Scholar]
  74. Santiago-Lopez, L.; Aguilar-Toal’a, J.E.; Hern’andez-Mendoza, A.; Vallejo-Cordoba, B.; Liceaga, A.; Gonz’alez-C’ordova, A.F. Invited review: Bioactive compounds produced during cheese ripening and health effects associated with aged cheese consumption. Int. J. Dairy Sci. 2018, 101, 3742–3757. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  75. Adesulu-Dahunsi, A.T.; Jeyaram, K.; Sanni, A.I. Probiotic and technological properties of exopolysaccharide producing lactic acid bacteria isolated from cereal based Nigerian fermented food products. Food Control. 2018, 92, 225–231. [Google Scholar] [CrossRef] [Green Version]
  76. Mapelli-Brahm, P.; Barba, F.J.; Remize, F.; Garcia, C.; Fessard, A.; Mousavi Khaneghah, A.; Mel’endez-Martínez, A.J. The impact of fermentation processes on the production, retention and bioavailability of carotenoids: An overview. Trends Food Sci. Technol. 2020, 99, 389–401. [Google Scholar] [CrossRef]
  77. Adebo, O.A.; Gabriela Medina-Meza, I. Impact of fermentation on the phenolic compounds and antioxidant activity of whole cereal grains: A mini review. Molecules 2020, 25, 927. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  78. Hiippala, K.; Jouhten, H.; Ronkainen, A.; Hartikainen, A.; Kailunanen, V.; Jalanka, J.; Satokari, R. The potential of gut commensals in reinforcing intestinal barrier function and alleviating inflammation. Nutrients 2018, 10, 988. [Google Scholar] [CrossRef] [Green Version]
  79. Wang, L.W.; Tancredi, D.J.; Thomas, D.W. The prevalence of gastrointestinal problems in children across the United States with autism spectrum disorders from families with multiple affected members. J. Dev. Behav. Pediatr. 2011, 32, 351–360. [Google Scholar] [CrossRef]
  80. Heidari, F.; Abbaszadeh, S.; Mirak, S.E.M. Evaluation Effect of Combination Probiotics and Antibiotics in the Prevention of Recurrent Urinary Tract Infection (UTI) in Women. Biomed. Pharmacol. J. 2017, 10, 691–698. [Google Scholar] [CrossRef]
  81. Selhub, E.M.; Logan, A.C.; Bested, A.C. Fermented foods, microbiota, and mental health: Ancient practice meets nutritional psychiatry. J. Physiol. Anthropol. 2014, 33, 2. [Google Scholar] [CrossRef] [Green Version]
  82. Lehtoranta, L.; Pitkaranta, A.; Korpela, R. Probiotics in respiratory virus infections. Eur. J. Clin. Microbiol. 2014, 33, 1289–1302. [Google Scholar] [CrossRef]
  83. Björkström, N.K.; Strunz, B.; Ljunggren, H.G. Natural killer cells in antiviral immunity. Nat. Rev. Immunol. 2022, 22, 112–123. [Google Scholar] [CrossRef]
  84. Guillemard, E.; Tanguy, J.; Flavigny, A.L.; De la Motte, S.; Schrezenmeir, J. Effects of consumption of a fermented dairy product containing the probiotic Lactobacillus casei DN-114 001 on common respiratory and gastrointestinal infections in shift workers in a randomized controlled trial. J. Am. Coll. Nutr. 2010, 29, 455–468. [Google Scholar] [CrossRef] [PubMed]
  85. Fujita, R.; Iimuro, S.; Shinozaki, T.; Sakamaki, K.; Uemura, Y.; Takeuchi, A.; Matsuyama, Y.; Ohashi, Y. Decreased duration of acute upper respiratory tract infections with daily intake of fermented milk: A multicenter, double-blinded, randomized comparative study in users of day care facilities for the elderly population. Am. J. Infect. Control 2013, 41, 1231–1235. [Google Scholar] [CrossRef] [PubMed]
  86. Araujo, G.V.; Oliveira Junior, M.H.; Peixoto, D.M.; Sarinho, E.S. Probiotics for the treatment of upper and lower respiratory-tract infections in children: Systematic review based on randomized clinical trials. J. Pediatr. 2015, 91, 413–427. [Google Scholar] [CrossRef]
  87. Wang, Y.; Li, X.; Ge, T.; Xiao, Y.; Liao, Y.; Cui, Y.; Zhang, Y.; Ho, W.; Yu, G.; Zhang, T. Probiotics for prevention and treatment of respiratory tract infections in children: A systematic review and meta-analysis of randomized controlled trials. Medicine 2016, 95, e4509. [Google Scholar] [CrossRef] [PubMed]
  88. Pu, F.; Guo, Y.; Li, M.; Zhu, H.; Wang, S.; Shen, X.; He, M.; Huang, C.; He, F. Yogurt supplemented with probiotics can protect the healthy elderly from respiratory infections: A randomized controlled open-label trial. Clin. Interv. Aging 2017, 12, 1223–1231. [Google Scholar] [CrossRef] [Green Version]
  89. Makino, S.; Ikegami, S.; Kume, A.; Horiuchi, H.; Sasaki, H.; Orii, N. Reducing the risk of infection in the elderly by dietary intake of yoghurt fermented with Lactobacillus delbrueckii ssp. bulgaricus OLL1073R-1. Br. J. Nutr. 2010, 104, 998–1006. [Google Scholar] [CrossRef] [Green Version]
  90. Yamamoto, Y.; Saruta, J.; Takahashi, T.; To, M.; Shimizu, T.; Hayashi, T.; Morozumi, T.; Kubota, N.; Kamata, Y.; Makino, S.; et al. Effect of ingesting yogurt fermented with Lactobacillus delbrueckii ssp. Bulgaricus OLL1073R-1 on influenza virus-bound salivary IgA in elderly residents of nursing homes: A randomized controlled trial. Acta Odontol. Scand. 2019, 77, 517–524. [Google Scholar] [CrossRef]
  91. Gouda, A.S.; Adbelruhman, F.G.; Sabbah Alenezi, H.; Mégarbane, B. Theoretical benefits of yogurt-derived bioactive peptides and probiotics in COVID-19 patients—A narrative review and hypotheses. Saudi J. Biol. Sci. 2021, 28, 5897–5905. [Google Scholar] [CrossRef]
  92. Choi, H.J.; Song, J.H.; Park, K.S.; Baek, S.; Lee, E.; Kwon, D. Antiviral activity of yogurt against enterovirus 71 in vero cells. Food Sci. Biotechnol. 2010, 19, 289–295. [Google Scholar] [CrossRef]
  93. Olaya Galán, N.N.; Ulloa Rubiano, J.C.; Velez Reyes, F.A.; Fernandez Duarte, K.P.; Salas Cárdenas, S.P.; Gutierrez Fernandez, M.F. In vitro antiviral activity of Lactobacillus casei and Bifidobacterium adolescentis against rotavirus infection monitored by NSP4 protein production. J. Appl. Microbiol. 2016, 120, 1041–1051. [Google Scholar] [CrossRef] [Green Version]
  94. Starosila, D.; Rybalko, S.; Varbanetz, L.; Ivanskaya, N.; Sorokulova, I. Anti-influenza Activity of a Bacillus subtilis Probiotic Strain. Antimicrob. Agents Chemother. 2017, 61, e00539-e17. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  95. Eguchi, K.; Fujitani, N.; Nakagawa, H.; Miyazaki, T. Prevention of respiratory syncytial virus infection with probiotic lactic acid bacterium Lactobacillus gasseri SBT2055. Sci. Rep. 2019, 9, 4812. [Google Scholar] [CrossRef]
  96. Das, G.; Heredia, J.B.; Pereira, M.L.; Coy-Barrera, E.; Oliveira, S.M.R.; Gutiérrez-Grijalva, E.P.; Cabanillas-Bojórquez, L.A.; Shin, H.; Patra, J.K. Korean traditional foods as antiviral and respiratory disease prevention and treatments: A detailed review. Trends Food Sci. Technol. 2021, 116, 415–433. [Google Scholar] [CrossRef] [PubMed]
  97. Park, M.K.; Ngo, V.; Kwon, Y.M.; Lee, Y.T.; Yoo, S.; Cho, Y.H.; Hong, S.M.; Hwang, H.S.; Ko, E.J.; Jung, Y.J.; et al. Lactobacillus plantarum DK119 as a probiotic confers protection against influenza virus by modulating innate immunity. PLoS ONE 2013, 8, e75368. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  98. Ögel, Z.; Öztürk, H. Antiviral mechanisms related to lactic acid bacteria and fermented food products. Biotech. Studies 2020, 29, 18–28. [Google Scholar]
  99. Rather, I.A.; Choi, K.H.; Bajpai, V.K.; Park, Y.H. Antiviral mode of action of Lactobacillus plantarum YML009 on Influenza virus H1N1. Bangladesh J. Pharmacol. 2015, 10, 475–482. [Google Scholar]
  100. Sugimura, T.; Takahashi, H.; Jounai, K.; Ohshio, K.; Kanayama, M.; Tazumi, K.; Tanihata, Y.; Miura, Y.; Fujiwara, D.; Yamamoto, N. Effects of oral intake of plasmacytoid dendritic cells-stimulative lactic acid bacterial strain on pathogenesis of influenza-like illness and immunological response to influenza virus. Br. J. Nutr. 2015, 114, 727–733. [Google Scholar] [CrossRef] [Green Version]
  101. Kawashima, T.; Hayashi, K.; Kosaka, A.; Kawashima, M.; Igarashi, T.; Tsutsui, H.; Tsuji, N.M.; Nishimura, I.; Hayashi, T.; Obata, A. Lactobacillus plantarum strain YU from fermented foods activates Th1 and protective immune responses. Int. Immunopharmacol. 2011, 11, 2017–2024. [Google Scholar] [CrossRef] [PubMed]
  102. Nagai, T.; Makino, S.; Ikegami, S.; Itoh, H.; Yamada, H. Effects of oral administration of yogurt fermented with Lactobacillus delbrueckii ssp. bulgaricus OLL1073R-1 and its exopolysaccharides against influenza virus infection in mice. Int. Immunopharmacol. 2011, 11, 2246–2250. [Google Scholar] [CrossRef]
  103. Bae, J.Y.; Kim, J.I.; Park, S.; Yoo, K.; Kim, I.H.; Joo, W.; Ryu, B.H.; Park, M.S.; Lee, I.; Park, M.S. Effects of Lactobacillus plantarum and Leuconostoc mesenteroides Probiotics on Human Seasonal and Avian Influenza Viruses. J. Microbiol. Biotechnol. 2018, 28, 893–901. [Google Scholar] [CrossRef] [Green Version]
  104. Glück, U.; Gebbers, J.O. Ingested probiotics reduce nasal colonization with pathogenic bacteria (Staphylococcus aureus, Streptococcus pneumoniae, and β-hemolytic streptococci). Am. J. Clin. Nutr. 2003, 77, 517–520. [Google Scholar] [CrossRef] [Green Version]
  105. Guillemard, E.; Tondu, F.; Lacoin, F.; Schrezenmeir, J. Consumption of a fermented dairy product containing the probiotic Lactobacillus casei DN-114001 reduces the duration of respiratory infections in the elderly in a randomised controlled trial. Br. J. Nutr. 2010, 103, 58–68. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  106. Merenstein, D.; Murphy, M.; Fokar, A.; Hernandez, R.K.; Park, H.; Nsouli, H.; Sanders, M.E.; Davis, B.A.; Niborski, V.; Tondu, F.; et al. Use of a fermented dairy probiotic drink containing Lactobacillus casei (DN-114 001) to decrease the rate of illness in kids: The DRINK study A patient-oriented, double-blind, cluster-randomized, placebo-controlled, clinical trial. Eur. J. Clin. Nutr. 2010, 64, 669–677. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  107. Corsello, G.; Carta, M.; Marinello, R.; Picca, M.; De Marco, G.; Micillo, M.; Ferrara, D.; Vigneri, P.; Cecere, G.; Ferri, P.; et al. Preventive effect of cow’s milk fermented with Lactobacillus paracasei CBA L74 on common infectious diseases in children: A multicenter randomized controlled trial. Nutrients 2017, 9, 669. [Google Scholar] [CrossRef] [PubMed]
  108. Nocerino, R.; Paparo, L.; Terrin, G.; Pezzella, V.; Amoroso, A.; Cosenza, L.; Cecere, G.; De Marco, G.; Micillo, M.; Albano, F.; et al. Cow’s milk and rice fermented with Lactobacillus paracasei CBA L74 prevent infectious diseases in children: A randomized controlled trial. Clin. Nutr. 2017, 36, 118–125. [Google Scholar] [CrossRef]
  109. Shida, K.; Sato, T.; Iizuka, R.; Hoshi, R.; Watanabe, O.; Igarashi, T.; Miyazaki, K.; Nanno, M.; Ishikawa, F. Daily intake of fermented milk with Lactobacillus casei strain Shirota reduces the incidence and duration of upper respiratory tract infections in healthy middle-aged office workers. Eur. J. Nutr. 2017, 56, 45–53. [Google Scholar] [CrossRef] [Green Version]
  110. Mai, T.T.; Thi Thu, P.; Thi Hang, H.; Trang, T.; Yui, S.; Shigehisa, A.; Tien, V.T.; Dung, T.V.; Nga, P.B.; Hung, N.T.; et al. Efficacy of probiotics on digestive disorders and acute respiratory infections: A controlled clinical trial in young Vietnamese children. Eur. J. Clin. Nutr. 2020, 75, 513–520. [Google Scholar] [CrossRef]
Table 1. The reported beneficial effect of FFs against respiratory tract infection.
Table 1. The reported beneficial effect of FFs against respiratory tract infection.
S. No. Type of StudyFermented ProductActive MicrobesSubjects/
Model
Key FindingsReference
1Randomized, double-blind, and controlled trial.Fermented milk containing probioticL. rhamnosus GGHealthy children↓ Risk of URTIs
↓ Duration of infection
[40]
2Randomized, double-blind, and controlled trial.fermented dairy drink containing probiotic
(Actimel®)
Probiotic: L. casei DN-114 001;
Conventional cultures: L. delbrueckii subsp. bulgaricus and S. thermophilus
Shift-based workers↓ Incidence of CIDs
↑ Neutrophil, leukocyte, and NK cell count and activity
[84]
3Multicenter, randomized, double-blind, placebo-controlled trial.Fermented
milk containing probiotic
L. casei strain ShirotaElderly
subjects
↓ Duration of URTIs[85]
4Randomized,
double-blind and placebo-controlled trial.
Fermented
yoghurt containing probiotic
Probiotic: L. delbrueckii
subsp. bulgaricus OLL1073R-1;
starter culture: S. thermophilus
Elderly subjects↑ NK cell activity
↓ Risk of the common cold
[89]
5Open, prospective trialFermented milk containing probioticsLactobacillus GG (ATCC 53103), Bifidobacterium
sp. B420, Lactobacillus acidophilus 145, and
S. thermophilus
Patients carrying potentially pathogenic bacteria in their nasal cavity↓ Nasal pathogenic bacteria
(↓ Gram +ve bacteria)
[104]
6Randomized, double-blind, and controlled trial.Fermented dairy drink containing probiotic
(Actimel®)
Probiotic: L. casei DN-114 001;
starter cultures: L. delbrueckii subsp. bulgaricus and S. thermophilus
Elderly subjects↓ Incidence of CIDs
↓Durations and episodes of URTIs
[105]
7Randomized, double-blind, and controlled trial.Fermented dairy drink containing probiotic
(Actimel®)
L. casei DN-114 001;
Starter cultures: L. delbrueckii subsp. bulgaricus and S. thermophilus
Healthy children ↓ Incidence of CIDs[106]
8Randomized, double-blind, and controlled trial.Fermented cow’s milkL. paracasei CBA L74 (heat-killed)Healthy children↓ Acute gastroenteritis
↓ URTIs
[107]
9Randomized, double-blind, and controlled trial.Fermented cow’s milk and fermented rice productL. paracasei CBA L74 (heat-killed)Healthy children↓ Incidence of CIDs
↓ URTIs; ↑ immunity
[108]
10Randomized, double-blind, and controlled trial.Fermented milk containing probioticL. casei strain
Shirota
Healthy office workers↓ Incidence and symptoms of URTIs
↑ Salivary cortisol levels in both probiotic and control group
[109]
11Controlled filed trialFermented milk containing probioticL. casei strain
Shirota
Children↓ Constipation
↓ Incidence of diarrhea
↓ Acute respiratory infection
[110]
CIDs: Respiratory and gastrointestinal common infectious diseases; NK: Natural killer; URTIs: Upper respiratory tract infections.
Publisher’s Note: MDPI stays neutral with regard to jurisdictional claims in published maps and institutional affiliations.

Share and Cite

MDPI and ACS Style

Kesika, P.; Thangaleela, S.; Sivamaruthi, B.S.; Bharathi, M.; Chaiyasut, C. Fermented Foods and Their Role in Respiratory Health: A Mini-Review. Fermentation 2022, 8, 162. https://doi.org/10.3390/fermentation8040162

AMA Style

Kesika P, Thangaleela S, Sivamaruthi BS, Bharathi M, Chaiyasut C. Fermented Foods and Their Role in Respiratory Health: A Mini-Review. Fermentation. 2022; 8(4):162. https://doi.org/10.3390/fermentation8040162

Chicago/Turabian Style

Kesika, Periyanaina, Subramanian Thangaleela, Bhagavathi Sundaram Sivamaruthi, Muruganantham Bharathi, and Chaiyavat Chaiyasut. 2022. "Fermented Foods and Their Role in Respiratory Health: A Mini-Review" Fermentation 8, no. 4: 162. https://doi.org/10.3390/fermentation8040162

Note that from the first issue of 2016, this journal uses article numbers instead of page numbers. See further details here.

Article Metrics

Back to TopTop